DOI QR코드

DOI QR Code

The Changes of Expression of Survivin by Butyrate in HCT116 Colon Cancer Cells

HCT116 대장암 세포주에서 Butyrate에 의한 Survivin 발현 변화

Park, Dong-Guk
박동국

  • Published : 20091100

Abstract

Purpose: The causes of colon cancer can be divided into genetic and environmental components. A high-fiber diet is known to reduce the risk of colon cancer. Dietary fiber is converted to short chain fatty acid, butyrate, in the colon by bacteria. Butyrate is used as an energy source for the colonic epithelial cells, and is known to induce apoptosis in colon cancer cell lines. Survivin, a recently discovered member of the IAP (inhibitor of apoptosis) family, is known to suppress apoptosis. Not only does it suppress cell apoptosis, but it also has a protective effect from disabling G2/M phase of the cell cycle by attaching to the microtubule of the mitotic spindle. The purpose of this study is to evaluate the effect of butyrate on the expression of survivin, in HCT116 colon cancer cell lines. Methods: Cytotoxicity of butyrate was measured by MTS method. Cell cycle phase and apoptosis was analyzed by flowcytometry. Protein expression of survivin was evaluated by Western blot analysis, and the mRNA expression by RT-PCR. Results: Butyrate can induce apoptosis in HCT116 colon cancer cell line at a concentration of 6 mM. Butyrate suppressed the expression of survivin mRNA and also the expression of cytosolic and nuclear survivin. In flowcytometric analysis, the apoptotic portion was increased and the proportions of S and M phase were decreased when cultured with butyrate. Conclusion: We concluded that butyrate could induce cellular apoptosis partially by suppressing the expression of survivin in HCT116 colon cancer cells.

Keywords

References

  1. Hague A, Manning AM, Hanlon KA, Huschtscha LI, Hart D, Paraskeva C. Sodium butyrate induces apoptosis in human colonic tumour cell lines in a p53-independent pathway: implications for the possible role of dietary fibre in the prevention of large-bowel cancer. Int J Cancer 1993;55:498-505 https://doi.org/10.1002/ijc.2910550329
  2. Bonnotte B, Favre N, Reveneau S, Micheau O, Droin N, Garrido C, et al. Cancer cell sensitization to fas-mediated apoptosis by sodium butyrate. Cell Death Differ 1998;5:480-7 https://doi.org/10.1038/sj.cdd.4400371
  3. Giardina C, Boulares H, Inan MS. NSAIDs and butyrate sensitize a human colorectal cancer cell line to TNF-alpha and Fas ligation: the role of reactive oxygen species. Biochim Biophys Acta 1999;1448:425-38 https://doi.org/10.1016/S0167-4889(98)00156-6
  4. McMillan L, Butcher SK, Pongracz J, Lord JM. Opposing effects of butyrate and bile acids on apoptosis of human colon adenoma cells: differential activation of PKC and MAP kinases. Br J Cancer 2003;88:748-53 https://doi.org/10.1038/sj.bjc.6600793
  5. Sandler RS, Lyles CM, Peipins LA, McAuliffe CA, Woosley JT, Kupper LL. Diet and risk of colorectal adenomas: macronutrients, cholesterol, and fiber. J Natl Cancer Inst 1993;85: 884-91 https://doi.org/10.1093/jnci/85.11.884
  6. Radley S, lmray CHE, Davis A, Hendrickse CW, Donovan IA, Lawson AM, et al. Duodenal bile acid profiles in patients with colorectal cancer or polyps. Eur J Gastroenterol Hepatol 1993;5:721-30 https://doi.org/10.1097/00042737-199309000-00010
  7. Bayerdorffer E, Mannes GA, Richter WO, Ochsenkuhn T, Wiebecke B, Kopcke W, et al. Increased serum deoxycholic acid levels in men with colorectal adenomas. Gastroenterology 1993;104:145-51 https://doi.org/10.1016/0016-5085(93)90846-5
  8. Mahmoud NN, Dannenberg AJ, Bilinski RT, Mestre JR, Chadburn A, Churchill M, et al. Administration of an unconjugated bile acid increases duodenal tumors in a murine model of familial adenomatous polyposis. Carcinogenesis 1999;20: 299-303 https://doi.org/10.1093/carcin/20.2.299
  9. Narisawa T, Magadia NE, Weisburger JH, Wynder EL. Promoting effect of bile acids on colon carcinogenesis after intrarectal instillation of N-methyl-N'-nitro-N-nitrosoguanidine in rats. J Natl Cancer Inst 1974;53:1093-7 https://doi.org/10.1093/jnci/53.4.1093
  10. Reddy BS, Wynder EL. Large-bowel carcinogenesis: fecal constituents of populations with diverse incidence rates of colon cancer. J Natl Cancer Inst 1973;50:1437-42 https://doi.org/10.1093/jnci/50.6.1437
  11. Sutherland LAM, Bird RP. The effect of chenodeoxycholic acid on the development of aberrant crypt foci in the rat colon. Cancer Lett 1994;76:101-7 https://doi.org/10.1016/0304-3835(94)90384-0
  12. DeRubertis FR, Craven PA, Saito R. Bile salt stimulation of colonic epithelial proliferation. Evidence for involvement of lipoxygenase products. J Clin Invest 1984;74:1614-24 https://doi.org/10.1172/JCI111577
  13. Martinez-Diez MC, Serrano MA, Monte MJ, Marin JJ. Comparison of the effects of bile acids on cell viability and DNA synthesis by rat hepatocytes in primary culture. Biochim Biophys Acta 2000;1500:153-60 https://doi.org/10.1016/S0925-4439(99)00099-X
  14. Cummings JH. Short chain fatty acids in the human colon. Gut 1981;22:763-79 https://doi.org/10.1136/gut.22.9.763
  15. Kruh J. Effects of sodium butyrate, a new pharmacological agent, on cells in culture. Mol Cell Biochem 1982;42:65-82
  16. Tsao D, Shi ZR, Wong A, Kim YS. Effect of sodium butyrate on carcinoembryonic antigen production by human colonic adenocarcinoma cells in culture. Cancer Res 1983;43:1217-22
  17. Boffa LC, Lupton JR, Mariani MR, Ceppi M, Newmark HL, Scalmati A, et al. Modulation of colonic epithelial cell proliferation, histone acetylation, and luminal short chain fatty acids by variation of dietary fiber (wheat bran) in rats. Cancer Res 1992;52:5906-12
  18. Barnard JA, Warwick G. Butyrate rapidly induces growth inhibition and differentiation in HT-29 cells. Cell Growth Differ 1993;4:495-501
  19. Berry RD, Paraskeva C. Expression of carcinoembryonic antigen by adenoma and carcinoma derived epithelial cell lines: possible marker of tumour progression and modulation of expression by sodium butyrate. Carcinogenesis 1988;9:447-50 https://doi.org/10.1093/carcin/9.3.447
  20. Rothe M, Pan MG, Henzel WJ, Ayres TM, Goeddel DV. The TNFR2-TRAF signaling complex contains two novel proteins related to baculoviral inhibitor of apoptosis proteins. Cell 1995;83:1243-52 https://doi.org/10.1016/0092-8674(95)90149-3
  21. Ambrosini G, Adida C, Altieri DC. A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med 1997;3:917-21 https://doi.org/10.1038/nm0897-917
  22. Gianani R, Jarboe E, Orlicky D, Frost M, Bobak J, Lehner R, et al. Expression of survivin in normal, hyperplastic, and neoplastic colonic mucosa. Hum Pathol 2001;32:119-25 https://doi.org/10.1053/hupa.2001.21897
  23. Chantalat L, Skoufias DA, Kleman JP, Jung B, Dideberg O, Margolis RL. Crystal structure of human survivin reveals a bow tie-shaped dimer with two unusual alpha-helical extensions. Mol Cell 2000;6:183-9 https://doi.org/10.1016/S1097-2765(00)00019-8
  24. Li F, Altieri DC. Transcriptional analysis of human survivin gene expression. Biochem J 1999;344:305-11 https://doi.org/10.1042/0264-6021:3440305
  25. Swana HS, Grossman D, Anthony JN, Weiss RM, Altieri DC. Tumor content of the antiapoptosis molecule survivin and recurrence of bladder cancer. N Engl J Med 1999;341:452-3 https://doi.org/10.1056/NEJM199908053410614
  26. Lu CD, Altieri DC, Tanigawa N. Expression of a novel antiapoptosis gene, survivin, correlated with tumor cell apoptosis and p53 accumulation in gastric carcinomas. Cancer Res 1998;58:1808-12
  27. Okada E, Murai Y, Matsui K, Isizawa S, Cheng C, Masuda M, et al. Survivin expression in tumor cell nuclei is predictive of a favorable prognosis in gastric cancer patients. Cancer Lett 2001;163:109-16 https://doi.org/10.1016/S0304-3835(00)00677-7
  28. Yamamoto T, Manome Y, Nakamura M, Tanigawa N. Downregulation of survivin expression by induction of the effector cell protease receptor-1 reduces tumor growth potential and results in an increased sensitivity to anticancer agents in human colon cancer. Eur J Cancer 2002;38:2316-24 https://doi.org/10.1016/S0959-8049(02)00247-2
  29. Miller AA, Kurschel E, Osieka R, Schmidt CG. Clinical pharmacology of sodium butyrate in patients with acute leukemia. Eur J Cancer Clin Oncol 1987;23:1283-7 https://doi.org/10.1016/0277-5379(87)90109-X

Cited by

  1. 삼지구엽초의 인체 대장암세포 HCT116 사멸 효과 vol.18, pp.2, 2009, https://doi.org/10.13050/foodengprog.2014.18.2.154
  2. 인간 대장암 HT-29 세포에서 제주조릿대의 세포사멸 효과 vol.24, pp.9, 2014, https://doi.org/10.5352/jls.2014.24.9.1012
  3. AGS 인체 위암세포에서 황흑산에 의한 ROS 생성 및 caspase 활성 의존적 apoptosis 유발 vol.25, pp.11, 2009, https://doi.org/10.5352/jls.2015.25.11.1235
  4. Combination Treatment of p53-Null HL-60 cells with Histone Deacetylase Inhibitors and Chlorambucil Augments Apoptosis and Increases BCL6 and p21 Gene Expression vol.12, pp.1, 2009, https://doi.org/10.2174/1874467211666181010161836